Thyroid Cancer - Sharma, 2015

Thyroid Cancer • Author: Pramod K Sharma, MD; Chief Editor: Arlen D Meyers, MD, MBA Practice Essentials Thyroid cancers

Views 139 Downloads 1 File size 315KB

Report DMCA / Copyright

DOWNLOAD FILE

Recommend stories

Citation preview

Thyroid Cancer • Author: Pramod K Sharma, MD; Chief Editor: Arlen D Meyers, MD, MBA

Practice Essentials Thyroid cancers represent approximately 1% of new cancer diagnoses in the United States each year. Thyroid malignancies are divided into papillary carcinomas (80%), follicular carcinomas (10%), medullary thyroid carcinomas (5-10%), anaplastic carcinomas (1-2%), primary thyroid lymphomas (rare), and primary thyroid sarcomas (rare). Hürthle cell carcinoma is a rare thyroid malignancy that is often considered a variant of follicular carcinoma. Hürthle cell carcinomas account for 2-3% of all thyroid malignancies. They occur more commonly in women than in men and typically manifest in the fifth decade of life. The clinical presentation is similar to that of other thyroid malignancies. See the image below.

A monomorphous cell population of Hürthle cells arranged in loosely cohesive clusters and single cells. The cells are polyhedral and have abundant granular cytoplasm with well-defined cell borders. The nuclei are enlarged and have a central prominent macronucleolus. For summarized information on staging and treatment, see Thyroid Cancer Staging and Thyroid Cancer Treatment Protocols.

Signs and symptoms Thyroid carcinoma most commonly manifests as a painless, palpable, solitary thyroid nodule. Patients or clinicians discover most of these nodules during routine palpation of the neck. Signs and symptoms associated with malignancy in thyroid nodules include the following: • Solitary nodules: Most likely to be malignant in patients older than 60 years and in patients

• • • • •

younger than 30 years Increased rate of malignancy in males Nodular growth Rapid growth: Ominous sign Usually painless (nontender to palpation); sudden onset of pain more strongly associated with benign disease (eg, hemorrhage into a benign cyst, subacute viral thyroiditis) Hard and fixed nodules

Diagnosis The key to the workup of the solitary thyroid nodule is to differentiate malignant from benign disease and, thus, to determine which patients require intervention and which patients may be monitored serially. History taking, physical examination, laboratory evaluation, and fine-needle aspiration biopsy (FNAB) are the mainstays in the evaluation of thyroid nodules. Imaging studies can be adjuncts in select cases. Examination in patients suspected of thyroid cancer includes the following: • Thorough head and neck examination, including thyroid gland and cervical soft tissues • Indirect laryngoscopy Firm cervical masses are highly suggestive of regional lymph node metastases. Vocal fold paralysis implies involvement of the recurrent laryngeal nerve. Procedures FNAB is the most important diagnostic tool in evaluating thyroid nodules and should be the first intervention. The following are the 4 possible results from this procedure: • • • •

Benign disease Malignant disease Indeterminate for diagnosis Nondiagnostic

Up to 50% of repeated biopsies result in a definitive diagnosis. Patients whose findings are indeterminate or nondiagnostic despite repeat biopsy can undergo surgery for lobectomy for tissue diagnosis. Nondiagnostic cases can also be monitored clinically, and radioiodine scans can be useful for determining the functional status of the nodule, because most hyperfunctioning nodules are benign. Laboratory testing The following laboratory studies may be used to assess patients with suspected thyroid cancer: • Serum thyroid-stimulating hormone concentration: Sensitive for hyperthyroidism/hypothyroidism and for evaluation of solitary thyroid nodules • Serum calcitonin/pentagastrin-stimulated calcitonin levels: Elevated levels highly suggestive of medullary thyroid carcinoma • Polymerase chain reaction (PCR) assay for germline mutations in the RET proto-oncogene: For diagnosis of familial medullary thyroid carcinoma Imaging studies The following imaging studies may be used to evaluate patients with suspected thyroid cancer: • Neck ultrasonography: Most common modality to evaluate thyroid disease; however, limited

usefulness for distinguishing between malignant and benign nodules • Thyroid radioiodine imaging: To determine functional status of a nodule but cannot exclude carcinoma • Neck computed tomography (CT) scanning or magnetic resonance imaging (avoid iodinated contrast agents): To evaluate soft-tissue extension of large or suspicious thyroid masses into the neck, trachea, or esophagus, and to assess metastases to the cervical lymph nodes; no role in routine management of solitary thyroid nodules A 2015 consensus statement from the American Thyroid Association on preoperative imaging for thyroid cancer surgery stated the following[1, 2] : • Ultrasonography remains the most important imaging modality in the evaluation of thyroid cancer and should be used routinely to assess the primary tumor and all associated cervical lymph node basins preoperatively • Ultrasonographically guided fine-needle aspiration of suspicious lymph nodes may be useful in guiding the extent of surgery • Cross-sectional imaging (CT scanning with contrast or magnetic resonance imaging [MRI]) may be considered in select circumstances to better characterize tumor invasion and bulky, inferiorly located, or posteriorly located lymph nodes; it may also be used when ultrasonographic expertise is not available

Management Malignant diagnoses require surgical intervention. Papillary thyroid carcinoma and medullary thyroid carcinoma are often positively identified on the basis of FNAB results alone. Cervical metastases discovered preoperatively or intraoperatively should be removed by means of en bloc lymphatic dissection of the respective cervical compartment (selective neck dissection) while sparing the nonlymphatic structures. Well-differentiated neoplasms Patients with follicular neoplasm, as determined with FNAB results, should undergo surgery for thyroid lobectomy for tissue diagnosis. The extent of surgical therapy for well-differentiated neoplasms is controversial. Primary treatment for papillary and follicular carcinoma is surgical excision whenever possible. Total thyroidectomy has been the mainstay for treating well-differentiated thyroid carcinoma. Modifications to total thyroidectomy include subtotal thyroidectomy to reduce the risk of recurrent laryngeal nerve injury and hypoparathyroidism. A 2015 consensus statement from the American Thyroid Association on the management of patients with differentiated thyroid cancer who have recurrent/persistent nodal disease stated the following[1, 3] : • The appropriate management of patients with nodal metastases may involve compartmental lymph-node dissection, active surveillance, radioactive iodine ablation therapy, external-beam radiation therapy, and/or nonsurgical, image-guided, minimally invasive ablative approaches • Biologic considerations include aggressive histology, extrathyroidal extension of primary tumor, and molecular prognosis for aggressive biology • Surgical/technical considerations include prior recurrences in the same or different compartments Hürthle cell carcinomas For patients with Hürthle cell carcinomas based on initial FNAB findings, most surgeons advocate an

aggressive approach with lobectomy and isthmectomy, followed by completion thyroidectomy with confirmation on final pathologic result. For tumors larger than 5 cm or for palpable lymphatic metastases, total thyroidectomy (including neck dissection for palpable lymph nodes) is often performed during the initial operation. Medullary thyroid carcinomas and familialmedullary thyroid carcinomas Sporadic medullary thyroid carcinomas and familial medullary thyroid carcinomas are treated with total thyroidectomy and lymphatic dissection of the anterior compartment of the neck. If the vasculature of the parathyroid gland is disrupted, autotransplantation of the parathyroid gland into the sternocleidomastoid muscle or the nondominant forearm is performed. In children with multiple endocrine neoplasia (MEN) type 2A and MEN 2B syndromes, prophylactic thyroidectomy and centralcompartment lymph-node dissection is performed. Anaplastic thyroid carcinoma, primary thyroid lymphoma, thyroid sarcoma Total or subtotal thyroidectomy is performed for anaplastic thyroid carcinoma when the extent of the tumor permits it. Tracheotomy is needed in cases with airway compromise due to tracheal invasion. Stage IE lymphomas may be treated with total thyroidectomy followed by postoperative radiation therapy. Surgical excision should not be performed if local infiltration into surrounding tissues is evident. Stage IIE lymphomas are treated with combined chemotherapy and radiation therapy. Doxorubicin or CHOP (ie, cyclophosphamide, hydroxydaunomycin, Oncovin [vincristine], prednisone) is the commonly used chemotherapeutic regimen. The treatment for thyroid sarcomas is total thyroidectomy. Radiation therapy may be used in an adjunctive setting. Postsurgical management After total thyroidectomy, patients undergo radioiodine scanning to detect regional or distant metastatic disease, followed by radioablation of any residual disease found. In addition, patients are given thyroid replacement therapy with T4 or triiodothyronine (T3). In patients with anaplastic thyroid carcinoma, chemotherapy and radiation therapy are typically administered in combination. Postoperative external-beam irradiation is effective in improving local control; this may also be used as primary treatment in unresectable cases. Chemotherapy (most commonly doxorubicin) is added for palliation. Next Section: Overview

Thyroid malignancy occurs with relative infrequency in the United States, although benign thyroid disease is relatively common. Although patients with thyroid cancers generally have a favorable prognosis compared with that of patients with many other solid tumors, an estimated 1200 patients died of thyroid cancer in the United States in 1998. Contemporary treatment of patients with thyroid malignancy requires a multidisciplinary approach involving an endocrinologist, a thyroid surgeon, a radiologist, and, on occasion, medical and radiation oncologists. For excellent patient education resources, visit eMedicineHealth's Thyroid and Metabolism Center. Also, see eMedicineHealth's patient education article Thyroid Problems. An image of Hurthle cells can be seen below. A monomorphous cell population of Hürthle cells arranged in loosely cohesive clusters and single cells. The cells are polyhedral and have abundant granular cytoplasm with well-defined cell borders. The nuclei are enlarged and have a central prominent macronucleolus.

Frequency Thyroid cancers represent approximately 1% of new cancer diagnoses each year. Approximately 23,500 cases of thyroid cancer are diagnosed yearly in the United States. The incidence of the disease is 3 times higher in women than in men; a study by Weir et al, from the US Centers for Disease Control and Prevention (CDC), predicted that by 2020, the largest increases in the annual number of cancer cases in women will be for cancers of the lung, breast, uterus, and thyroid.[4] The incidence of thyroid cancer peaks in the third and fourth decades of life. Thyroid cancers are divided into papillary carcinomas, follicular carcinomas, medullary thyroid carcinomas (MTCs), anaplastic carcinomas, primary thyroid lymphomas, and primary thyroid sarcomas. Papillary carcinoma represents 80% of all thyroid neoplasms. Follicular carcinoma is the second most common thyroid cancer, accounting for approximately 10% of cases. MTCs represent 510% of neoplasms. Anaplastic carcinomas account for 1-2%. Primary lymphomas and sarcomas are rare. Using data on 497 US counties from the National Cancer Institute's Surveillance, Epidemiolgy, and End Results program, Morris et al found that the tripling of the incidence of papillary thyroid cancer in the past 3 decades is directly correlated with demographic and age-based markers of access to health care, suggesting widespread overdiagnosis of the disease. Supporting this conclusion, rates of mortality from thyroid cancer remained stable during this period.[5, 6]

Etiology Thyroid carcinomas arise from the 2 cell types present in the thyroid gland. The endodermally derived follicular cell gives rise to papillary, follicular, and probably anaplastic carcinomas. The neuroendocrine-derived calcitonin-producing C cell gives rise to MTCs. Thyroid lymphomas arise from intrathyroid lymphoid tissue, whereas sarcomas likely arise from connective tissue in the thyroid gland. Radiation exposure significantly increases the risk for thyroid malignancies, particularly papillary thyroid carcinoma. This finding was observed in children exposed to radiation after the nuclear bombings in Hiroshima and Nagasaki during World War II. Additional evidence was gathered after atomic bombs were tested in the Marshall Islands, after the accident at the Chernobyl nuclear power plant, and in patients who received low-dose radiation therapy for benign disorders (eg, acne,

adenotonsillar hypertrophy). Low-dose radiation exposure from imaging studies has not been found to have a tumorigenic effect. Radiation targeting the thyroid gland (eg, iodine-131 ablation of the thyroid) or high-dose external-beam radiation therapy does not appear to increase the risk of papillary thyroid carcinoma. This is presumably because cell killing increases with these doses. Low dietary intake of iodine does not increase the incidence of thyroid cancers overall. However, populations with low dietary iodine intake have a high proportion of follicular and anaplastic carcinomas. Previous Next Section: Presentation

Clinical Presentation History Thyroid carcinoma most commonly manifests as a painless, palpable, solitary thyroid nodule. Patients or physicians discover most of these nodules during routine palpation of the neck. Palpable thyroid nodules are present in approximately 4-7% of the general population, and most represent benign disease. High-resolution ultrasonography reportedly depicts thyroid nodules in 19-67% of randomly selected individuals. An estimated 5-10% of solitary thyroid nodules are malignant. Palpable and nonpalpable nodules of similar size have the same risk of malignancy. The patient's age at presentation is important because solitary nodules are most likely to be malignant in patients older than 60 years and in patients younger than 30 years. In addition, thyroid nodules are associated with an increased rate of malignancy in male individuals. Growth of a nodule may suggest malignancy. Rapid growth is an ominous sign. Malignant thyroid nodules are usually painless. Sudden onset of pain is more strongly associated with benign disease, such as hemorrhage into a benign cyst or subacute viral thyroiditis, than with malignancy. Hoarseness suggests involvement of the recurrent laryngeal nerve and vocal fold paralysis. Dysphagia may be a sign of impingement of the digestive tract. Heat intolerance and palpitations suggest autonomously functioning nodules. Medullary carcinoma can occur as part of multiple endocrine neoplasia (MEN) 2A or 2B syndrome, as well as familial MTC (FMTC) syndrome. Patients with a family history of thyroid cancer should be evaluated with vigilance.

Physical examination Physical examination should include thorough head and neck examination with careful attention to the thyroid gland and cervical soft tissues, as well as indirect laryngoscopy. Solitary thyroid nodules can vary from soft to hard. Hard and fixed nodules are more suggestive of malignancy than supple mobile nodules are. Thyroid carcinoma is usually nontender to palpation. Firm cervical masses are highly suggestive of regional lymph node metastases. Vocal fold paralysis implies involvement of the recurrent laryngeal nerve. Previous Next Section: Evaluation and Management of the Solitary Thyroid Nodule

Evaluation and Management of the Solitary Thyroid Nodule The key to the workup of the solitary thyroid nodule is to differentiate malignant from benign disease and, thus, to determine which patients require intervention and which patients may be monitored serially. History taking, physical examination, laboratory evaluation, and fine-needle aspiration biopsy (FNAB) are the mainstays in the evaluation of thyroid nodules. Imaging studies can be adjuncts in select cases. A 2015 consensus statement from the American Thyroid Association on preoperative imaging for thyroid cancer surgery stated the following[1, 2] : • Ultrasonography remains the most important imaging modality in the evaluation of thyroid cancer and should be used routinely to assess the primary tumor and all associated cervical lymph node basins preoperatively; positive lymph nodes may be distinguished from normal nodes based on size, shape, echogenicity, hypervascularity, loss of hilar architecture, and the presence of calcifications • Ultrasonographically guided fine-needle aspiration of suspicious lymph nodes may be useful in guiding the extent of surgery • Cross-sectional imaging (CT scanning with contrast or magnetic resonance imaging [MRI]) may be considered in select circumstances to better characterize tumor invasion and bulky, inferiorly located, or posteriorly located lymph nodes; it may also be used when ultrasonographic expertise is not available • The above recommendations are applicable to initial and revision surgery; functional imaging with positron emission tomography (PET) or PET-CT scanning may be helpful in cases of recurrent cancer with positive tumor markers and negative anatomic imaging

Fine-needle aspiration biopsy FNAB is the most important diagnostic tool in evaluating thyroid nodules and should be the first intervention. The technique is inexpensive and easy to perform, and it causes few complications. To perform FNAB, comfortably position both the patient and the physician. Extend the patient's neck slightly and palpate the nodule with the nondominant hand. Clean the skin with alcohol and infiltrate the area with local anesthesia. Place a 21- to 25-gauge needle on the end of a syringe. Many physicians use trigger-style aspirating handles on the syringe. Introduce 2 mL of air into the syringe, and place the needle into the skin. Apply negative pressure to the syringe, and pass the needle through the nodule, which is identified by using the nondominant hand. After several passes, release the negative pressure, and withdraw the needle. Use the air remaining in the syringe to expel the specimen from the hub and needle onto a glass slide or into cytologic solution for a cell block. Fix the slide in alcohol for Papanicolaou and hematoxylin-eosin staining. Some slides can be air dried and stained with Romanowsky stain (Diff-Quick). Successful diagnosis by the cytologist depends on accurate sampling of the nodule and specimen cellularity. For this reason, several authors recommend performing at least 3 aspirations to ensure adequacy of the specimen and to minimize false-negative results. Ultrasonographic guidance can help to increase the accuracy of FNAB. Danese et al report increased false-negative rates with palpation FNAB compared with ultrasonography-guided FNAB. The 4 results from FNAB are benign disease, malignant disease, indeterminate for diagnosis, and nondiagnostic. In their review of several large series, Gharib and Goellner (1993) found that 69% of

FNAB results were benign, 4% were malignant, 10% were indeterminate, and 17% were nondiagnostic.[7] Their false-positive rate was 2.9%, and their false-negative rate was 5.2%. Sensitivity and specificity were 83% and 92%, respectively. Results of FNAB determine the next step in managing the thyroid nodule. A definitive diagnosis is obtained in as many as 50% of repeated biopsies. Patients whose findings are nondiagnostic despite repeat biopsy can undergo surgery for lobectomy for tissue diagnosis, or they can be monitored clinically. In these circumstances, radioiodine scans can be useful for determining the functional status of the nodule, as most hyperfunctioning nodules are benign. Indeterminate biopsy findings are labeled suspicious at some institutions. When cellular material is adequate for evaluation but when malignant and benign disease cannot be differentiated, biopsy results can be labeled suspicious. Patients with a suspicious diagnosis should undergo lobectomy for definitive diagnosis. Malignant diagnoses require surgical intervention. Papillary thyroid carcinoma and MTC are often positively identified on the basis of FNAB results alone. In patients with these carcinomas, definitive surgical planning can be undertaken at the outset. However, it is nearly impossible to distinguish a follicular adenoma from a follicular carcinoma on the basis of FNAB findings. Patients with follicular neoplasm, as determined with FNAB results, should undergo surgery for thyroid lobectomy for tissue diagnosis. These patients require complete thyroidectomy if a malignancy is discovered on review of the pathology. Some controversy exists regarding the extent of thyroidectomy (total thyroidectomy, subtotal thyroidectomy, or lobectomy) for a particular pathologic diagnosis. Each pathologic diagnosis and its corresponding extent of thyroidectomy are discussed below. Complications of FNAB are few and generally minor. The most common complications are minor hematoma, ecchymosis, and local discomfort. Clinically significant hematoma and swelling is exceedingly rare. Inadvertent puncture of the trachea, carotid artery, or jugular vein usually does not cause clinically significant problems and is managed with the application of local pressure.

Laboratory evaluation The serum thyroid-stimulating hormone (TSH) concentration is a highly sensitive measure for hyperthyroidism or hypothyroidism. A sensitive TSH assay is useful in the evaluation of solitary thyroid nodules. A low serum TSH value suggests an autonomously functioning nodule, which typically is benign. However, malignant disease cannot be ruled out on the basis of low or high TSH levels. Other thyroid function tests are usually not necessary in the initial workup. Serum thyroglobulin measurements are not helpful diagnostically because they are elevated in most benign thyroid conditions. Elevated serum calcitonin levels are highly suggestive of MTC. Serum calcitonin measurement, which was once the mainstay in the diagnosis of FMTC, has been replaced by sensitive polymerase chain reaction (PCR) assays for germline mutations in the RET proto-oncogene. These mutations are present in patients with MEN 2A, MEN 2B, and FMTC (see Genetic testing for MEN and FMTC in the Medullary Thyroid Carcinoma section). However, calcitonin and the more sensitive pentagastrinstimulated calcitonin are used as tumor markers to monitor patients who have been treated for MTC. Because of the low incidence of MTC overall, testing of serum calcitonin is not a cost-effective screening tool in the primary workup of thyroid nodules.

Imaging procedures Ultrasonography is the imaging modality most commonly used to evaluate thyroid disease. This noninvasive study enables accurate evaluation of the thyroid gland. However, the usefulness of ultrasonography for distinguish between malignant and benign nodules is limited. Simple cysts found on sonograms are benign, but simple cysts are rarely found. Cysts are most commonly complex, with at least some solid component that could potentially harbor malignancy. Microcalcifications noted on sonograms are associated with thyroid malignancy. Ultrasonography is highly sensitive for thyroid nodules and can depict nodules only a few millimeters in size. A sonogram ordered to evaluate a solitary nodule often reveals additional nodules of questionable clinical significance. The accuracy of FNAB results increases when sonographic guidance is used. Use of ultrasonography-guided FNAB can be useful for biopsy of small or difficult-to-palpate thyroid nodules as well as for FNAB of nodules in children. Ultrasonography can also be useful for accurate measurement of thyroid nodules that are being monitored serially. In 2013, researchers at the Mayo Clinic reported that recent dramatic increases in the diagnosis of lowrisk thyroid cancer in the United States are fuelled by the overuse of ultrasonography.[8, 9] The incidence of thyroid cancer in the United States has tripled in the past 3 decades, from 3.6 per 100,000 in 1973 to 11.6 per 100,000 in 2009, making it one of the fastest growing diagnoses. The vast majority of the thyroid tumors being detected are small low-risk papillary thyroid cancers that are unlikely to ever progress enough to cause symptoms or death. That this represents overdiagnosis is supported by the observation that the death rate for these cancers has remained stable (0.5 per 100,000 in 1979 and in 2009), even with the increasing incidence.[8, 9] Radioiodine imaging can help in determining the functional status of a nodule. Nonfunctional nodules do not take up radiolabeled iodine-123 and appear as cold spots in the thyroid (cold nodules).[10] Hyperfunctioning nodules take up radioiodine and appear as hot spots (hot nodules). Warm nodules appear similar to the surrounding normal thyroid tissue. Hot or warm nodules were historically thought to be benign; therefore, they did not require further evaluation for malignancy. However, in a review of 5000 patients undergoing thyroidectomy regardless of radioimaging findings, Ashcraft and Van Herle (1981) found that 4% of hot nodules harbored malignancy. Carcinoma cannot be excluded on the basis of radioiodine scans. Therefore, radioiodine scans are usually not helpful for the routine evaluation of thyroid nodules. In select situations, radioiodine studies can be diagnostic adjuncts. When results of repeated FNAB of a nodule are nondiagnostic, a radioiodine imaging can help in directing management if a hot nodule is to be monitored clinically. CT scanning and MRI can be used to evaluate soft-tissue extension of large or suspicious thyroid masses into the neck, trachea, or esophagus and to assess metastases to the cervical lymph nodes. These studies do not have a role in the routine management of solitary thyroid nodules. The use of iodinated contrast agents should be avoided in patients with possible thyroid carcinoma because they interfere with the postoperative use of radioactive iodine. Previous Next Section: Well-Differentiated Thyroid Carcinoma

Well-Differentiated Thyroid Carcinoma A 2015 consensus statement from the American Thyroid Association on the management of patients with differentiated thyroid cancer who have recurrent/persistent nodal disease stated the following[1, 3] : • The appropriate management of patients with nodal metastases may involve compartmental lymph-node dissection, active surveillance (watchful waiting with serial cervical ultrasonographic evaluations), radioactive iodine ablation therapy, external-beam radiation therapy, and/or nonsurgical, image-guided, minimally invasive ablative approaches • Biologic considerations include aggressive histology, extrathyroidal extension of primary tumor, and molecular prognosis for aggressive biology • Surgical/technical considerations include prior recurrences in the same or different compartments

Papillary carcinoma Clinical features Papillary carcinoma is the most common thyroid malignancy, representing approximately 80%. Papillary carcinoma and follicular carcinoma make up the well-differentiated thyroid carcinomas. Women develop papillary cancer 3 times more frequently than men do, and the mean age at presentation is 34-40 years. Cases can occur familially, either alone or in association with Gardner syndrome (familial adenomatous polyposis). As noted above, radiation exposure, especially during childhood, is associated with the development of papillary thyroid carcinoma. Tumors typically appear after a latency period of about 10-20 years. In addition, an increased incidence of papillary cancer is hypothesized among patients with Hashimoto thyroiditis (chronic lymphocytic thyroiditis). Despite this possibility, the rate of malignancy for a given nodule in people with Hashimoto thyroiditis is similar to that of individuals with a normal gland. Papillary carcinoma is a slow-growing tumor that arises from the thyroxine (T4)- and thyroglobulinproducing follicular cells of the thyroid. The cells are TSH sensitive and take up iodine. They produce thyroglobulin in response to TSH stimulation. This feature has both diagnostic and therapeutic value for managing residual disease and recurrences after surgical excision (see Treatment and Prognosis below). Pathology On gross pathologic examination, papillary carcinomas are whitish invasive neoplasms with ill-defined margins. Under microscopy, the tumors are unencapsulated neoplasms that characteristically grow with papillae consisting of neoplastic epithelium overlying fibrovascular stalks. Very differentiated tumors can have a complex arborizing pattern. Nuclei have an empty ground-glass appearance with characteristic nuclear grooves and pseudoinclusions. Mitoses are rare. Another histologic feature is the presence of psammoma bodies, which occur in 50% of papillary carcinomas. Psammoma bodies are calcific concretions that have a circular laminated appearance. They are found in the stroma of the tumor. In addition, many papillary carcinomas contain areas that show a follicular growth pattern. However, when the nuclear features in follicular areas are the same as those in papillary areas, the tumor behaves like a classic papillary carcinoma and should be designated as such. Papillary carcinoma may be multicentric, with foci present in both the ipsilateral and contralateral lobes.

Local invasion Tumors can grow directly through the thyroid capsule to invade surrounding structures. Growth into the trachea can occur, producing hemoptysis. Extensive involvement can cause airway obstruction. The recurrent laryngeal nerves can become involved because of their proximity in the tracheoesophageal groove. Patients present with a hoarse, breathy voice and, occasionally, dysphagia. Regional and metastatic disease Another common feature of papillary carcinoma is its propensity to spread to the cervical lymph nodes. Clinically evident lymph node metastases are present in approximately one third of patients at presentation. Microscopic metastases are present in one half. The most common site of lymph node involvement is in the central compartment (level 6) located medial to the carotid sheaths on both sides, with extension from the hyoid bone superiorly to the sternal notch inferiorly. The jugular lymph node chains (levels 2-4) are the next most common sites of cervical node involvement. Lymph nodes in the posterior triangle of the neck (level 5) may also develop metastases. This finding has important implications on the treatment algorithm for patients in this situation (see Treatment and Prognosis below and the images below).

Algorithm for the management of a solitary thyroid nodule. FNAB = fine needle aspiration biopsy; US = ultrasonography.

Algorithm for the management of malignant thyroid neoplasms. FNAB = fine needle aspiration biopsy; XRT = external-beam radiation therapy. A study by Kim et al indicated that in patients with papillary thyroid cancer, a tumor size of greater than 1 cm and nonretroesophageal lateral lymph node metastasis are independent predictors of right retroesophageal lymph node metastasis.[11] Approximately 5-10% of patients with papillary thyroid carcinoma develop distant metastases. Distant spread of papillary carcinoma typically affects the lungs and bone.

Follicular carcinoma Clinical features Follicular carcinoma is the second most common thyroid malignancy and represents about 10% of thyroid cancers. Follicular carcinoma represents an increased portion of thyroid cancers in regions where dietary intake of iodine is low. Similar to papillary carcinoma, follicular carcinoma occurs 3 times more frequently in women than in men. Patients with follicular carcinoma are typically older than those with papillary carcinoma at presents. The mean age range at diagnosis is late in the fourth to sixth decades. Like papillary carcinomas, follicular carcinomas arise from the follicular cells of the thyroid. The neoplastic cells are TSH sensitive as well, taking up iodine and producing thyroglobulin—a feature that is exploited diagnostically and therapeutically (see Postoperative radioiodine scanning and ablation

below). Pathology On gross pathology, the tumors appear as round, encapsulated, light brown neoplasms. Fibrosis, hemorrhage, and cystic changes are found in the lesions. Under microscopy, the tumors contain neoplastic follicular cells, which overall can have a solid, trabecular, or follicular growth pattern (that usually produces microfollicles). The follicular cells in these tumors do not have characteristic features like papillary carcinoma cells. Follicular carcinomas are differentiated from benign follicular adenomas by tumor capsule invasion and/or vascular invasion. For this reason, differentiating follicular adenomas from follicular carcinomas is extremely difficult with FNAB cytology and frozen section analysis. The tumors are divided into minimally invasive and widely invasive lesions depending on the histologic evidence of capsule and vascular invasion. Immunohistochemical staining for thyroglobulin and cytokeratins is nearly always positive. Local invasion Local invasion can occur as it does with papillary carcinoma, with the same presenting features (see Local invasion for Papillary Carcinoma, above). Cervical and distant metastases Unlike papillary carcinoma, cervical metastases from follicular carcinomas are uncommon. However, the rate of distant metastasis is significantly increased (approximately 20%). Lung and bone are the most common sites.

Surgical treatment The extent of surgical therapy for well-differentiated neoplasms is controversial. Primary treatment for papillary and follicular carcinoma is surgical excision whenever possible. Total thyroidectomy has been the mainstay for treating well-differentiated thyroid carcinoma. In this procedure, all apparent thyroid tissue is surgically removed. Major complications in this procedure are recurrent laryngeal nerve injury and hypoparathyroidism from inadvertent damage or removal of the parathyroid glands. Complications associated with total thyroidectomy are discussed in the Technique of Thyroidectomy section below. After total thyroidectomy, patients undergo radioiodine scanning to detect regional or distant metastatic disease (see Postoperative radioiodine scanning and ablation below), followed by radioablation of any residual disease found. Over the years, modifications to total thyroidectomy have been proposed in an effort to reduce recurrent laryngeal nerve injury and hypoparathyroidism associated with total thyroidectomy. Subtotal thyroidectomy has been proffered as an alternative to total thyroidectomy. With subtotal thyroidectomy, a small portion of gross thyroid tissue opposite the side of malignancy is left in place to minimize the risk of injuring the recurrent laryngeal nerve and of inadvertently removing the parathyroid glands on that side. Patients usually receive postoperative radioiodine treatment in an attempt to ablate the remaining thyroid tissue. With improved stratification of patients into prognostic groups (see Prognostic factors below), some surgeons have proposed thyroid lobectomy with isthmectomy alone as definitive treatment for patients at low risk for recurrent or metastatic disease. This approach remains to be substantiated as a feasible alternative to total thyroidectomy. Using the Surveillance, Epidemiology, and End Results (SEER) database, one study compared the

overall survival (OS) and cause-specific survival (CSS) of 23,605 subjects with papillary or follicular thyroid cancer treated with local excision, lobectomy, near-total thyroidectomy, or total thyroidectomy. The 10-year OS and CSS results concluded that total thyroidectomy resulted in improved survival over other techniques; poorer outcomes were associated with age, stage T3/T4 disease, positive nodes, and tumor size.[12] According to a 2009 study by Asari et al, of 207 patients with follicular thyroid carcinoma, the 127 patients with minimum growth had no lymph node metastases. The authors state that thyroidectomy is still recommended for all patients with follicular thyroid carcinoma, although patients with widely invasive disease may need more aggressive surgical treatment. Patients with minimal disease invasion have an excellent prognosis with limited need for nodal surgery.[13]

Management of neck The neck must be thoroughly examined for lymphatic metastases. Ultrasonography of the neck with particular attention to the central compartment (level 6) is an effective diagnostic approach. FNAB of suspicious lymph nodes can be performed. Cervical metastases discovered preoperatively or intraoperatively should be removed by means of en bloc lymphatic dissection of the respective cervical compartment (selective neck dissection) while sparing the nonlymphatic structures. Excision of single nodes, known as berry picking, is inadequate therapy for metastatic disease. Elective neck dissection (removal of clinically benign neck lymphatic tissue) in a well-differentiated carcinoma is not indicated because postoperative radioiodine treatment effectively treats microscopic lymphatic metastases.

Postoperative radioiodine scanning and ablation Because differentiated thyroid tissue and well-differentiated thyroid carcinomas are TSH sensitive and because they take up iodine, radioiodine preferentially targets residual normal or malignant tissue after thyroidectomy. Therefore, radioiodine can be given in diagnostic doses to detect residual normal or neoplastic tissue in the body and in therapeutic doses to ablate this tissue. After thyroidectomy, use of radioiodine scanning and ablation has become commonplace for diagnosing and treating residual thyroid tissue, as well as regional and distant metastases from well-differentiated thyroid carcinomas. Pretherapeutic iodine-uptake scanning is controversial because of its cost and because of concerns about131 I-induced tumor stunning, which may decrease the effectiveness of radioiodine treatment. After thyroidectomy, patients are given thyroid replacement therapy with T4 (Synthroid) or triiodothyronine (T3, Cytomel).131 I or123 I scanning is performed when the patient is in a hypothyroid state (TSH >30-50). Approximately 4-6 weeks after thyroidectomy, hypothyroid can be induced by discontinuing replacement (T4 for 4 weeks or T3 for 2 weeks) to obtain high serum TSH levels. A diagnostic dose of131 I or123 I is given initially. Whole-body scanning is performed to detect any tissue taking up radioiodine. If any normal thyroid remnant or metastatic disease is detected, a therapeutic dose of131 I is administered to ablate the tissue. Posttreatment scanning should also be performed because it may reveal metastatic disease not otherwise noted. The role of recombinant human TSH (Thyrogen) in remnant ablation continues to evolve. Thyrogen is approved for postsurgical remnant ablation in Europe but not the United States. Barbaro et al found equivalent results in postsurgical remnant ablation when they compared traditional T4 withdrawal with the discontinuation of T4 1 day before TSH stimulation. Thyrogen stimulation avoids the discomfort of patients having to discontinue thyroid replacement and is especially useful in those unable to tolerate hypothyroidism or to generate a high TSH level.

If a treatment dose of131 I is required, diagnostic thyroid scanning is repeated while the patient is in the hypothyroid state about 6 months after initial treatment. Again, if the diagnostic scan is positive, an additional therapeutic dose is given. This process is repeated until the diagnostic scan is negative. A promising new development for follow-up thyroid scanning is the use of recombinant human TSH as opposed to withdrawing T4 to increase autogenous TSH levels. This approach avoids the discomfort of having to discontinue thyroid replacement therapy for these scans. A retrospective analysis of more than 1000 patients with papillary thyroid cancer who underwent total thyroidectomies found that most patients with low-risk local disease and some with high-risk T3 tumors who did not receive radioiodine remnant ablation after surgery had high 5-year recurrence-free survival rates. This suggests that physicians should carefully consider whether the benefits outweigh the risks associated with radioiodine remnant ablation when debating the possibility of employing this technique in individual patients.[14, 15] A study by Ruel et al indicated that adjuvant radioiodine therapy can improve the overall survival rate in patients with intermediate-risk papillary thyroid cancer. The study, which had a mean 6-year followup period, involved 21,870 adult patients who underwent total thyroidectomy for intermediate-risk papillary thyroid cancer, including 15,418 who received adjuvant radioiodine treatment and 6452 who did not. The investigators found that the risk of death was reduced by 29% in the radioiodine patients, with the risk decreased by 36% in those under age 45 years.[16]

Thyroid suppression After thyroidectomy and radioiodine ablation, patients with well-differentiated thyroid carcinoma are maintained on thyroid-suppression suppression. Patients take T4 in daily doses sufficient to suppress TSH production by the pituitary. Low TSH levels in the bloodstream reduce tumoral growth rates and reduce recurrence rates of well-differentiated thyroid carcinomas. The extent to which TSH should be suppressed is controversial. Most authors recommend reducing TSH levels to 0.1 mU/L. This level provides adequate thyroid suppression while avoiding deleterious cardiac and bone effects of profound thyroid suppression.

Follow-up care Patients are regularly monitored every 6-12 months with serial radioiodine scanning and serum thyroglobulin measurements after surgery and radioiodine therapy. Thyroglobulin is a useful marker of tumor recurrence because well-differentiated thyroid cancers synthesize thyroglobulin. However, it is useful only after total thyroid ablation. Serum thyroglobulin is measured at the time of follow-up thyroid scanning, during the withdrawal of thyroid hormone or the administration of recombinant TSH. Serum antithyroglobulin antibodies are measured in addition to thyroglobulin because their presence invalidates the assay. Thyroglobulin antibody levels should be obtained with each thyroglobulin measurement. Rising thyroglobulin level after thyroid ablation suggests recurrence. Ultrasonography of the neck can also be used to detect regional recurrences.

Pharmacologic therapy Sorafenib (Nexavar) was approved in November 2013 for differentiated thyroid cancer (DTC) that is refractory to radioactive iodine treatment. In a study of 417 patients with progressive radioiodinerefractory DTC, treatment with sorafenib, an orally active inhibitor of VEGFR1-3 and Raf kinases, significantly improved progression-free survival (10.8 months) compared with placebo (5.8 months).

[17, 18] Tumor

histology was 57% papillary, 25% follicular, and 10% poorly differentiated. The majority of the patients (96%) had metastatic disease, of which 71% of the target lesions were in the lung, 40% in lymph nodes, and 14% in bone. At the time of the report, median overall survival had not yet been reached in either study arm, and 70% of placebo patients had started open-label sorafenib.[17, 18] Thus, all reported responses were partial: 12.2% in the sorafenib group vs 0.5% in the placebo group. The rate of stable disease for 6 months or longer was 42% in the sorafenib group and 33% in the placebo group. A second oral VEGF inhibitor for refractory DTC was approved, in February 2015. Approval for lenvatinib (Lenvima) was based on the SELECT phase-3 trial. Compared with patients on placebo, those taking lenvatinib showed significant improvements in progression-free survival (median period of 18.3 months, compared with 3.6 months with placebo). The response rate among patients with iodine-131-refractory thyroid cancer was also significantly better with lenvatinib than with placebo.[19, 20]

Management of recurrence Recurrences are best treated with surgical excision if the disease is clinically evident and surgically accessible. Nonlocalized recurrences detected on the basis of elevated thyroglobulin levels are treated with131 I. On occasion, recurrent tumors do not concentrate iodine. Positron emission tomography (PET) may be helpful in localizing disease in such circumstances. When surgical excision of recurrent disease is not feasible, external-beam radiation therapy may be useful. Chemotherapy, usually with doxorubicin, is reserved for tumors that do no respond to other treatments and for palliative care. Response rates of 35-40% are reported, though complete responses to chemotherapy are rare.

Prognostic factors The long-term disease-free survival with aggressive treatment and management is nearly 90% overall. A variety of factors are associated with prognosis, as listed below. • Age: The patient's age at diagnosis is one of the most important prognostic features of welldifferentiated thyroid carcinoma. Cancer-related death is most likely to occur if the patient is >40 years at the time of diagnosis. Recurrences are most common in patients whose disease is diagnosed when they were < 20 years or >60 years. • Sex: Men are twice as likely as women to die from thyroid cancer. • Size: The size of the primary tumor is related to survival. Patients with primary tumors >4 cm have increased recurrence and cancer-related mortality rates. • Histology: Overall, papillary carcinoma is associated a 30-year cancer-related death rate of 6%. Follicular carcinoma has a 30-year cancer-related death rate of 15%. • Local invasion: Invasion of surrounding tissues outside of thyroid indicates biologic aggressiveness and significantly worsens the patient's prognosis. • Lymph node metastasis: Lymph node metastasis does not appear to be as important in the outcome of well-differentiated thyroid carcinomas as in the outcome of most other solid tumors. • Distant metastasis: Distant metastasis at initial examination is associated with a 68.1-fold increase in the rate of disease-specific death. Previous Next Section: Hürthle Cell Carcinomas

Hürthle Cell Carcinomas Clinical features Hürthle cell carcinoma is a rare thyroid malignancy that is often considered a variant of follicular carcinoma. Also known as oncocytic carcinoma, Hürthle cell carcinoma has unique biologic features. About 75-100% of the tumor is composed of Hürthle cells, which are also known as oxyphilic, oncocytic, Askanazy, or large cells. These are large, polygonal follicular cells that contain abundant granular acidophilic cytoplasm. Hürthle cells can be found in a variety of benign thyroid conditions, such as Hashimoto thyroiditis, Graves disease, and multinodular goiter. Benign neoplasms, called Hürthle cell adenomas, that contain more than 75% Hürthle cells can also occur. Hürthle cell carcinomas account for 2-3% of all thyroid malignancies. They occur more commonly in women than in men and typically manifest in the fifth decade of life. The clinical presentation is similar to that of other thyroid malignancies.

Pathology On pathologic examination, Hürthle cell carcinoma, like follicular carcinoma, is differentiated from Hürthle cell adenoma by the presence of capsular invasion, vascular invasion, or both. On gross evaluation, Hürthle cell carcinomas appear brown and solid. Most have an appreciable capsule. Under microscopy, the tumors have a solid or trabecular growth pattern of large, granular, polygonal Hürthle cells. Because malignant tumors are difficult to identify on the basis of cellular elements alone, Hürthle cell tumors identified on FNAB findings cannot be categorized as malignant or benign. Therefore, when FNAB results suggest a Hürthle cell neoplasm, a surgically obtained specimen is required.

Management Hürthle cell carcinomas behave aggressively. Patients with these lesions are at high risk for recurrent and metastatic disease. These tumors most often do not take up radioactive iodine, thereby removing the diagnostic and therapeutic benefits that papillary and follicular carcinomas have. Most surgeons advocate an aggressive approach to treating these tumors. Patients with a diagnosis of Hürthle cell neoplasm based on FNAB findings undergo lobectomy and isthmectomy. If, the final pathologic result confirm Hürthle cell carcinoma, patients return to surgery for completion thyroidectomy. For tumors >5 cm or for palpable lymphatic metastases, total thyroidectomy (including neck dissection for palpable lymph nodes) is often performed during the initial operation.

Prognosis Patients with Hürthle cell carcinoma should be monitored closely for recurrent and metastatic disease. The overall 5-year survival rate is 50-60%. Because tumors do not take up iodine and are not TSH sensitive, thyroid suppression and radioiodine therapy have little value. External-beam radiation therapy can used to treat metastatic disease. Surgery is the mainstay of treatment. Previous Next Section: Medullary Thyroid Carcinoma

Medullary Thyroid Carcinoma Clinical features MTCs represent approximately 5% of all thyroid malignancies. A slight female preponderance is observed. Tumors arise from the parafollicular C cells of the thyroid gland. C cells are neural-crest derivatives and produce calcitonin. About 75% of MTCs occur sporadically, and 25% occur familially. Familial cases are commonly multifocal throughout the thyroid gland, whereas sporadic cases are usually not multifocal. Patients may present with clinical evidence of MTC, or they may present before MTCs develop if they are from a family with known FMTC syndrome. New germline mutations can also occur. Patients with new germline mutations present with MTCs without a positive family history, but they are at risk for passing on the syndrome. The FMTC syndromes consist of MEN 2A, MEN 2B, and FMTC. They are inherited in an autosomal dominant fashion. Children inheriting an FMTC syndrome have a 100% risk of developing MTC. MEN 2A (Sipple syndrome) consists of MTC, pheochromocytoma (in 50% of patients), and hyperparathyroidism (10-20% of patients). MEN 2B consists of MTC, pheochromocytoma (in 50% of patients), marfanoid habitus, and ganglioneuromatosis. FMTC consists of MTC alone. MTC in MEN 2B has the most aggressive biologic features. In this situation, MTC usually develops by the age of 10 years, and it has a high propensity for rapid growth and metastasis. MTC in MEN 2A can appear in the first decade of life, and it almost always develops by the second decade. MTC in FMTC usually develops during adulthood.

Diagnosis of sporadic cases Sporadic cases typically manifest with painless solitary thyroid nodules, like other thyroid malignancies do. Likewise, symptoms of pain, dysphagia, and hoarseness can develop with local invasion.

Genetic testing for MEN and FMTC Genetic testing is now the mainstay in the diagnosis of the FMTC syndromes. RET proto-oncogene mutations (on chromosome arm 10q) have been discovered in each of the MTC syndromes. The RET proto-oncogene is a receptor tyrosine kinase whose exact function and role in these syndromes has not been elucidated. Patients with MEN 2A have germline RET mutations resulting in substitutions of conserved cysteine residues in exons 10 and 11. All patients with MEN 2B have a germline mutation resulting in a threonine-for-methionine substitution in codon 918 of exon 16. Mutations are described in exons 13 and 14 in patients with FMTC. Genetic screening with sensitive PCR assays for germline RET mutations is routinely performed in atrisk patients. Children of parents known to have MEN or FMTC are tested for RET mutations to guide therapy and future genetic counseling. In addition, patients presenting with sporadic MTC should undergo RET mutational analysis to rule out new spontaneous germline mutations, which should prompt the testing of offspring for similar mutations.

Biochemical testing for MTC Because MTC cells produce calcitonin, elevated serum calcitonin levels are diagnostic of MTC. Although routine measurement of serum calcitonin has low yield in managing the solitary thyroid nodule because of the uncommon nature of MTCs, it is useful in the surveillance of patients with a history of MTC and in managing familial forms. Stimulating calcitonin release by using intravenous pentagastrin increases the sensitivity of the test. For pentagastrin-stimulated calcitonin evaluation, a baseline plasma calcitonin level is measured, followed by the intravenous administration of pentagastrin 0.5 mg/kg and serial measurements of calcitonin 1.5 and 5 minutes after injection. Elevated basal or stimulated calcitonin levels above the normal range for the laboratory strongly suggest MTC. Plasma calcitonin levels are commonly increased before clinical evidence of MTC appears. Although this finding was once the mainstay in diagnosing familial forms of MTC, results of genetic testing have largely supplanted it. Plasma calcitonin testing is now used for the early detection of MTC in patients already known to be at risk for MTC because of their family history and genetic results. This level is most commonly used as a tumor marker to identify residual and metastatic disease after thyroidectomy to treat MTC.

Pathology On gross examination, MTCs are fairly well circumscribed, though they are unencapsulated. They are typically tannish pink and often contain yellow granular regions, which represent focal calcification. Most tumors arise in the middle and upper third of the thyroid lobes, commensurate with the location of the parafollicular C cells in the thyroid gland. Sporadic tumors are unilateral, and inherited forms usually involve both thyroid lobes. MTCs can have a varied microscopic appearance. The tumors typically have a lobular, trabecular, insular, or sheetlike growth pattern. Some tumors have a fibrotic character. Malignant cells may appear round, polygonal, or spindle shaped. The cytoplasm is eosinophilic and finely granular. In the stroma, characteristic deposits of amyloid are commonly observed. This amyloid has typical green birefringence on Congo red staining, and this is a feature unique to MTC among thyroid malignancies. Immunohistochemical stains for calcitonin and carcinoembryonic antigen are microscopically useful for differentiating MTC from other tumors. A unique feature to the familial cases of MTC is the finding of C-cell hyperplasia, which can help in distinguishing familial cases from sporadic ones. C-cell hyperplasia is considered a precursor to MTC and is usually adjacent to foci of MTC. The finding of C-cell hyperplasia with MTC should raise the suspicion for familial disease.

Treatment Both sporadic MTCs and FMTCs are treated with total thyroidectomy and lymphatic dissection of the anterior compartment of the neck (level VI). If the vasculature of the parathyroid gland is disrupted, autotransplantation of the parathyroid gland into the sternocleidomastoid muscle or the nondominant forearm is performed. Metastasis to the cervical lymph nodes is common in patients with MTC, particularly those with familial forms with multicentricity and bilaterality of the primary tumor. Lymph node metastases can occur in more than 50% of patients. Both before and at the time of surgery, the lateral jugular lymphatics should carefully be palpated for evidence of metastatic disease. Selective neck dissection (sparing nonlymphatic structures when possible) of levels II, III, IV, and V is performed when

metastases are clinically evident.

Prophylactic thyroidectomy in patients with MEN 2A and MEN 2B MTC is the most common cause of mortality in patients with MEN 2A and MEN 2B, and many patients who inherit these syndromes develop MTC in the first decade of life. Therefore, prophylactic thyroidectomy and central-compartment lymph-node dissection is being performed in children with these syndromes. Surgery is offered to patients when the diagnosis is made on the basis of RET mutational analysis. Children with RET mutations whose parents decline surgery should be monitored with annual measurement of calcitonin levels. Thyroidectomy is performed when results are abnormal.

Follow-up care After receiving treatment for MTC, patients are monitored with annual measurement of serum calcitonin levels for surveillance. Pentagastrin-stimulated calcitonin testing is no longer widely available. Carcinoembryonic antigen is another tumoral marker associated with the recurrence of MTC, and it may also be used for surveillance. Patients with elevated levels of calcitonin or carcinoembryonic antigen are evaluated for recurrent disease. Neck, abdominal, and pelvic CT or MRI may be used to detect disease if metastasis or recurrence is suspected. Ultrasonography may be useful to localize cervical disease. In addition, radionuclide studies and selective venous catheterization with sampling of calcitonin levels can be performed to localize recurrences. The role of PET is evolving. Radiation therapy is used in an adjuvant setting at some centers, and it can be used to treat patients with surgically inoperable recurrences and metastases. Because MTC does not concentrate iodine, radioiodine therapy has no role in follow-up care or treatment. A variety of chemotherapeutic regimens have been used to treat metastatic disease. MTC is relatively insensitive to chemotherapy, though partial responses have been obtained. To date, the most effective combination is dacarbazine, vincristine, and cyclophosphamide. Adding doxorubicin to this regimen, some investigators have gained a partial response rate of about 35%. Vandetanib (Caprelsa) and cabozantinib (Cometriq) are tyrosine kinase inhibitors approved by the FDA for progressive, metastatic medullary thyroid cancer. These agents target various tyrosine kinases including MET, RET, and VEGFR-2.

Prognosis The overall prognosis for patients with MTC is worse than that of patients with well-differentiated carcinoma. The reported 10-year survival rate is 65% overall. Young age, small primary tumor, low stage of disease, and completeness of initial resection improve survival. Patients with MEN 2B have a prognosis substantially worse than that of patients with MEN 2A, though the prognosis for both groups has improved with early diagnosis and intervention. Previous Next Section: Anaplastic Carcinoma and Other Thyroid Carcinomas

Anaplastic Carcinoma and Other Thyroid Carcinomas Anaplastic Thyroid Carcinoma Clinical features Anaplastic thyroid carcinoma is one of the least common thyroid carcinomas, accounting for 1.6% of all thyroid cancers. However, it has the most aggressive biologic behavior of all thyroid malignancies and one of the worst survival rates of all malignancies in general. Like papillary and follicular carcinomas, anaplastic thyroid carcinomas affect more women than men, with a female-to-male ratio of about 2-3:1. Patients with anaplastic thyroid carcinomas present later than those with other thyroid malignancies; the former most typically present in the sixth or seventh decade of life.[21] Anaplastic thyroid carcinoma manifests as a rapidly growing thyroid mass in contrast to welldifferentiated carcinomas, which are comparatively slow growing. Patients commonly present with associated symptoms due to local invasion. Hoarseness and dyspnea resulting from the involvement of the recurrent laryngeal nerve and airway occur in as many as 50% of patients. Physical examination reveals a firm thyroid mass or masses that are most often larger than 5 cm at presentation. About 30% of patients have vocal cord paralysis, and cervical metastases are palpable on examination in 40% of patients. At least one half of patients already have distant metastases at the time of diagnosis. The most common sites of involvement are the lungs, bones, and brain. Pathology On gross examination, anaplastic thyroid carcinoma is a large and invasive tumor. Areas of focal necrosis and hemorrhage may be present throughout the tumor, giving a highly variable appearance. The tumor often extends through the capsule of the thyroid gland itself. Areas of well-differentiated thyroid carcinoma are often found concomitantly, and anaplastic thyroid carcinoma is believed to arise from a preexisting, well-differentiated thyroid carcinoma. On microscopic evaluation, squamoid, spindle cell, and giant cell variants are observed. All 3 histologic variants show high mitotic activity, large foci of necrosis, and notable infiltration. Immunohistochemical stains are often positive for low-molecular-weight keratins and occasionally positive for thyroglobulin. Regarding their ultrastructure, the neoplasms have epithelial features (eg, desmosomes, tight junctions) that are helpful for differentiating them from sarcomas. Small cell carcinomas, which usually represent lymphomas, may be confused with anaplastic thyroid carcinoma. Treatment The progression of disease is rapid, and most patients die from local airway obstruction or complications of pulmonary metastases within 1 year despite all treatment efforts. Total or subtotal thyroidectomy is performed when the extent of the permits it. Neck dissection is added to manage palpable cervical metastases. Complete excision is often impossible because many patients present with clinically significant local extension. Tracheal and laryngeal resection is usually not performed to remove disease because of the poor prognosis in these circumstances. Tracheotomy is needed in cases with airway compromise due to tracheal invasion. External-beam irradiation is effective in improving local control. It is added postoperatively or used as primary treatment in unresectable cases. Chemotherapy is added for palliation. Doxorubicin is the most commonly used chemotherapeutic agent. Chemotherapy and radiation therapy typically administered used in combination. Prognosis

Anaplastic thyroid carcinoma is poorly responsive to multimodality therapy, and median survival is 8.1 months. Young age, unilateral tumors, small tumors (< 5 cm), no local invasion of the surrounding tissue, and a lack of cervical metastases are relatively favorable prognostic indicators. Patients with these features may have slightly prolonged survival. Long-term survival should prompt a reconsideration of the diagnosis of anaplastic thyroid carcinoma; the original tumor is usually found to be a variant of MTC or thyroid lymphoma.

Primary Thyroid Lymphoma Clinical features Primary lymphomas of the thyroid gland represent approximately 2-5% of all thyroid malignancies. Most thyroid lymphomas are non-Hodgkin B-cell tumors. The next most common histologic type is low-grade malignant lymphoma of mucosa-associated lymphoid tissue (MALT). Hodgkin lymphoma, Burkitt cell lymphoma, and T-cell lymphoma have also been reported. The incidence peaks in the sixth decade of life, and most diagnoses are made in patients aged 50-80 years. Women are more commonly affected than men, with a female-to-male ratio of 4:1. This tumor is highly associated with chronic lymphocytic thyroiditis (Hashimoto thyroiditis). Almost all patients with primary thyroid lymphoma have either a clinical history or histologic evidence of chronic lymphocytic thyroiditis. The risk of primary thyroid lymphoma increases 70-fold in patients with chronic lymphocytic thyroiditis compared with the general population. The most common clinical presentation is an enlarging thyroid mass. Patients may have clinical or serologic evidence of hypothyroidism. Local extension into the aerodigestive tract or surrounding tissues may cause dysphagia, dyspnea, or symptoms of pressure in the neck. Vocal fold paralysis and hoarseness suggest involvement of the recurrent laryngeal nerve. Regional and distant lymphadenopathy is common. Diagnosis is based on the patient's clinical presentation with a positive tissue diagnosis. FNAB may be useful for diagnosing thyroid lymphoma, but it is considered less reliable with this lesion than with other thyroid malignancies. Lymphoma may be difficult to differentiate from chronic lymphocytic thyroiditis. Surgical biopsy of the lesion is preferred for diagnosing thyroid lymphoma. Biopsy enables thorough histochemical and immunohistochemical analysis to confirm the diagnosis. Tumor cells are positive for leukocyte-common antigen and for B- or T-cell markers depending on the type of tumor. Staging of thyroid lymphomas is important for therapeutic and prognostic purposes. Staging involves CT scanning of the brain, neck, chest, abdomen, and pelvis, as well as bone marrow biopsy. Most primary thyroid lymphomas are localized to the thyroid gland alone and, therefore, classified as stage IE (localized to an extranodal site). Involved regional lymph nodes increase the stage to IIE. Treatment and prognosis Stage IE lymphomas may be treated with total thyroidectomy followed by postoperative radiation therapy. Surgical excision should not be performed if local infiltration into surrounding tissues is evident. Stage IIE lymphomas are treated with combined chemotherapy and radiation therapy. Doxorubicin or CHOP (ie, cyclophosphamide, hydroxydaunomycin, Oncovin [vincristine], prednisone) is the commonly used chemotherapeutic regimen. Most thyroid lymphomas are stage IE, which have a 5-year survival rate of up to 85%. Spread beyond the thyroid gland reduces the 5-year survival rate to about 35%. Lymphomas at stages higher than this worsen the prognosis.

Sarcoma of the Thyroid Gland Sarcomas that arise in the thyroid gland are uncommon. They are aggressive tumors that most likely arise from stromal or vascular tissue in the gland. Malignancies that appear to be sarcomas should be differentiated from anaplastic thyroid carcinomas, which can appear sarcomatous. The treatment for thyroid sarcomas is total thyroidectomy. Radiation therapy may be used in an adjunctive setting. Most sarcomas are unresponsive to chemotherapy. Recurrence is common, as it is with sarcomas arising in other sites in the body, and the patient's overall prognosis is poor. Previous Next Section: Technique of Thyroidectomy

Technique of Thyroidectomy Thyroid surgery is performed to diagnose or treat thyroid disease. The extent of surgery ranges from isthmectomy alone (for small nodules truly localized to the isthmus) to subtotal thyroidectomy, total thyroidectomy, or extended thyroidectomy. Radioiodine studies performed after total thyroidectomy usually show residual normal thyroid tissue despite total thyroidectomy. Principles of thyroid surgery are accurate execution of the planned excision, avoidance of injury to the recurrent laryngeal nerve, avoidance of injury to or devascularization of the parathyroid glands, and meticulous hemostasis.

Indications • Indications for thyroid lobectomy • Biopsy of a solitary thyroid nodule suggestive of malignancy • Management of compressive or cosmetic symptoms due to a multinodular goiter • Management of a well-differentiated thyroid carcinoma in a low-risk patient (This is controversial.) • Indications for total thyroidectomy • Management of a well-differentiated thyroid malignancy • Management of an MTC • Management of a sarcoma of the thyroid gland • Management of stage IE thyroid lymphoma • Management of an obstructive goiter (Consider subtotal thyroidectomy.)

Preoperative considerations Vocal fold mobility should always be determined before thyroid surgery. If lobectomy for biopsy is planned, discuss the potential need for completion thyroidectomy with the patient.

Operative technique Positioning of the patient is important. Place the patient in a supine position with his or her neck extended by using a shoulder roll. Plan a horizontal incision in a natural skin crease to contour the curvature of the neck. The location should overly the thyroid gland, evenly extending between the anterior aspect of the sternocleidomastoid muscles on both sides. Elevate skin flaps superiorly and inferiorly in a subplatysmal plane. (Platysma is often absent in the midline.) Ligate the anterior jugular veins only if they directly limit exposure. Separate the sternohyoid and sternothyroid muscles in the median raphe, and retract them laterally to expose the cricoid cartilage and thyroid isthmus. In the anterior region, dissect the strap muscles off the face of the thyroid lobe (bilaterally for total thyroidectomy). Supracapsular dissection is continued until the superior pole and its vascular pedicle are isolated. The superior pole vessels are divided and ligated. The dissection continues laterally with division of the middle thyroid vein. The thyroid lobe is gradually medialized. Careful blunt dissection is performed to identify the recurrent laryngeal nerve in the tracheoesophageal groove. After the recurrent nerve is identified, carefully follow the nerve superiorly toward the larynx. The

nerve passes closely to the Berry ligament, but its position varies. After the nerve is thoroughly identified in this region, divide the ligament to release the thyroid gland. Keep the location of the parathyroid glands in mind during lateral dissection. Avoid disturbing the gland and vasculature as much as possible. Dissection close to the thyroid capsule minimizes this risk. If possible, ligate the inferior thyroid artery only after the vessels to the inferior parathyroid gland branch. If a parathyroid gland is inadvertently removed, reimplant it in the sternocleidomastoid muscle or on the volar surface of the forearm after slicing it into small pieces and marking it with a surgical clip. During superior dissection, remember the nearby location of the external branch of the superior laryngeal nerve, which innervates the cricothyroid muscle. Ligation of the superior-pole vessels tight to the thyroid in this area avoids inadvertent injury to this nerve. If only lobectomy is planned, divide the thyroid isthmus in the midline. Ligate the final soft-tissue attachments, and remove and label the lobe. Send it to the pathology laboratory. When total thyroidectomy is performed, the surgeon may elect not to divide the thyroid isthmus in the midline, but rather, to perform lateral dissection bilaterally. Identify the recurrent laryngeal nerves, and manage the inferior and superior vascular pedicles similarly. Remove the gland in 1 piece, label it, and send it for pathologic analysis. Irrigate the wound, and achieve meticulous hemostasis. The decision to place a passive or closed suction drain often depends on the surgeon's judgment. If a drain is used, place it into the wound and bring it out laterally through the incision or through a separate stab incision. The present authors have found the routine use of drains unnecessary. Reapproximate the sternothyroid and sternohyoid, and carefully close the skin in layers.

Postoperative care If a surgical drain is placed, maintain it until its output has diminished sufficiently, usually on the first postoperative day. Hypocalcemia may occur in patients who have undergone total thyroidectomy. Assess for hypocalcemia by inquiring about perioral paraesthesia. In a patient with hypocalcemia, tapping on preauricular region overlying the trunk of the facial nerve may cause ipsilateral contraction of the face (Chovstek sign). Measure ionized calcium postoperatively. Hypocalcemia may require calcium and vitamin D supplementation. Manage pain with acetaminophen and narcotics as needed. Previous

Sharma, Pramod K. 2015. Thyroid Cancer. http://emedicine.medscape.com/article/851968overview#aw2aab6b2